Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters











Publication year range
1.
RNA ; 27(9): 1046-1067, 2021 09.
Article in English | MEDLINE | ID: mdl-34162742

ABSTRACT

RNA exosomopathies, a growing family of diseases, are linked to missense mutations in genes encoding structural subunits of the evolutionarily conserved, 10-subunit exoribonuclease complex, the RNA exosome. This complex consists of a three-subunit cap, a six-subunit, barrel-shaped core, and a catalytic base subunit. While a number of mutations in RNA exosome genes cause pontocerebellar hypoplasia, mutations in the cap subunit gene EXOSC2 cause an apparently distinct clinical presentation that has been defined as a novel syndrome SHRF (short stature, hearing loss, retinitis pigmentosa, and distinctive facies). We generated the first in vivo model of the SHRF pathogenic amino acid substitutions using budding yeast by modeling pathogenic EXOSC2 missense mutations (p.Gly30Val and p.Gly198Asp) in the orthologous S. cerevisiae gene RRP4 The resulting rrp4 mutant cells show defects in cell growth and RNA exosome function. Consistent with altered RNA exosome function, we detect significant transcriptomic changes in both coding and noncoding RNAs in rrp4-G226D cells that model EXOSC2 p.Gly198Asp, suggesting defects in nuclear surveillance. Biochemical and genetic analyses suggest that the Rrp4 G226D variant subunit shows impaired interactions with key RNA exosome cofactors that modulate the function of the complex. These results provide the first in vivo evidence that pathogenic missense mutations present in EXOSC2 impair the function of the RNA exosome. This study also sets the stage to compare exosomopathy models to understand how defects in RNA exosome function underlie distinct pathologies.


Subject(s)
Exoribonucleases/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Mutation, Missense , RNA, Fungal/genetics , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Amino Acid Substitution , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Dwarfism/enzymology , Dwarfism/genetics , Dwarfism/pathology , Exoribonucleases/chemistry , Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/chemistry , Exosome Multienzyme Ribonuclease Complex/metabolism , Facies , Gene Expression , Glycine/chemistry , Glycine/metabolism , Hearing Loss/enzymology , Hearing Loss/genetics , Hearing Loss/pathology , Humans , Models, Biological , Models, Molecular , Protein Conformation , RNA, Fungal/chemistry , RNA, Fungal/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Retinitis Pigmentosa/enzymology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid , Syndrome
2.
Toxicol Lett ; 349: 115-123, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34089817

ABSTRACT

Cisplatin, the most widely used platinum-based anticancer drug, often causes progressive and irreversible sensorineural hearing loss in cancer patients. However, the precise mechanism underlying cisplatin-associated ototoxicity is still unclear. Nicotinamide adenine dinucleotide (NAD+), a co-substrate for the sirtuin family and PARPs, has emerged as a potent therapeutic molecular target in various diseases. In our investigates, we observed that NAD+ level was changed in the cochlear explants of mice treated with cisplatin. Supplementation of a specific inhibitor (TES-1025) of α-amino-ß-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD), a rate-limiting enzyme of NAD+de novo synthesis pathway, promoted SIRT1 activity, increased mtDNA contents and enhanced AMPK expression, thus significantly reducing hair cells loss and deformation. The protection was blocked by EX527, a specific SIRT1 inhibitor. Meanwhile, the use of NMN, a precursor of NAD+ salvage synthesis pathway, had shown beneficial effect on hair cell under cisplatin administration, effectively suppressing PARP1. In vivo experiments confirmed the hair cell protection of NAD+ modulators in cisplatin treated mice and zebrafish. In conclusion, we demonstrated that modulation of NAD+ biosynthesis via the de novo synthesis pathway and the salvage synthesis pathway could both prevent ototoxicity of cisplatin. These results suggested that direct modulation of cellular NAD+ levels could be a promising therapeutic approach for protection of hearing from cisplatin-induced ototoxicity.


Subject(s)
Enzyme Inhibitors/pharmacology , Hair Cells, Auditory/drug effects , Hearing Loss/prevention & control , Hearing/drug effects , NAD/biosynthesis , Ototoxicity/prevention & control , Sirtuin 1/metabolism , Animals , Animals, Genetically Modified , Carboxy-Lyases/antagonists & inhibitors , Carboxy-Lyases/metabolism , Cisplatin , Disease Models, Animal , Enzyme Activation , Hair Cells, Auditory/enzymology , Hair Cells, Auditory/pathology , Hearing Loss/chemically induced , Hearing Loss/enzymology , Hearing Loss/physiopathology , Lateral Line System/drug effects , Lateral Line System/enzymology , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/pathology , Ototoxicity/enzymology , Ototoxicity/etiology , Ototoxicity/physiopathology , Zebrafish
3.
Hear Res ; 388: 107896, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31982642

ABSTRACT

The development of knockin mice with Cre recombinase expressed under the control of the promoter for choline acetyltransferase (ChAT) has allowed experimental manipulation of cholinergic circuits. However, currently available ChATCre mouse lines are on the C57BL/6J strain background, which shows early onset age-related hearing loss attributed to the Cdh23753A mutation (a.k.a., the ahl mutation). To develop ChATCre mice without accelerated hearing loss, we backcrossed ChATIRES-Cre mice with CBA/CaJ mice that have normal hearing. We used genotyping to obtain mice homozygous for ChATIRES-Cre and the wild-type allele at the Cdh23 locus (ChATCre,Cdh23WT). In the new line, auditory brainstem response thresholds were ∼20 dB lower than those in 9 month old ChATIRES-Cre mice at all frequencies tested (4-31.5 kHz). These thresholds were stable throughout the period of testing (3-12 months of age). We then bred ChATCre,Cdh23WT animals with Ai14 reporter mice to confirm the expression pattern of ChATCre. In these mice, tdTomato-labeled cells were observed in all brainstem regions known to contain cholinergic cells. We then stained the tissue with a neuron-specific marker, NeuN, to determine whether Cre expression was limited to neurons. Across several brainstem nuclei (pontomesencephalic tegmentum, motor trigeminal and facial nuclei), 100% of the tdTomato-labeled cells were double-labeled with anti-NeuN (n = 1896 cells), indicating Cre-recombinase was limited to neurons. Almost all of these cells (1867/1896 = 98.5%) also stained with antibodies against ChAT, indicating that reporter label was expressed almost exclusively in cholinergic neurons. Finally, an average 88.7% of the ChAT+ cells in these nuclei were labeled with tdTomato, indicating that the Cre is expressed in a large proportion of the cholinergic cells in these nuclei. We conclude that the backcrossed ChATCre,Cdh23WT mouse line has normal hearing and expresses Cre recombinase almost exclusively in cholinergic neurons. This ChATCre,Cdh23WT mouse line may provide an opportunity to manipulate cholinergic circuits without the confound of accelerated hearing loss associated with the C57BL/6J background. Furthermore, comparison with lines that do show early hearing loss may provide insight into possible cholinergic roles in age-related hearing loss.


Subject(s)
Brain Stem/enzymology , Choline O-Acetyltransferase/metabolism , Cholinergic Fibers/enzymology , Hearing Loss/prevention & control , Hearing , Integrases/metabolism , Animals , Auditory Threshold , Brain Stem/physiopathology , Cadherins/genetics , Choline O-Acetyltransferase/genetics , Crosses, Genetic , DNA-Binding Proteins/metabolism , Evoked Potentials, Auditory, Brain Stem , Female , Gene Knock-In Techniques , Hearing Loss/enzymology , Hearing Loss/genetics , Hearing Loss/physiopathology , Integrases/genetics , Male , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Mutation , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic , Species Specificity
4.
Hum Mol Genet ; 28(24): 4103-4112, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31628468

ABSTRACT

ELMOD3, an ARL2 GTPase-activating protein, is implicated in causing hearing impairment in humans. However, the specific role of ELMOD3 in auditory function is still far from being elucidated. In the present study, we used the CRISPR/Cas9 technology to establish an Elmod3 knockout mice line in the C57BL/6 background (hereinafter referred to as Elmod3-/- mice) and investigated the role of Elmod3 in the cochlea and auditory function. Elmod3-/- mice started to exhibit hearing loss from 2 months of age, and the deafness progressed with aging, while the vestibular function of Elmod3-/- mice was normal. We also observed that Elmod3-/- mice showed thinning and receding hair cells in the organ of Corti and much lower expression of F-actin cytoskeleton in the cochlea compared with wild-type mice. The deafness associated with the mutation may be caused by cochlear hair cells dysfunction, which manifests with shortening and fusion of inner hair cells stereocilia and progressive degeneration of outer hair cells stereocilia. Our finding associates Elmod3 deficiencies with stereocilia dysmorphologies and reveals that they might play roles in the actin cytoskeleton dynamics in cochlear hair cells, and thus relate to hearing impairment.


Subject(s)
Deafness/enzymology , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/metabolism , Hearing Loss/enzymology , Stereocilia/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Cochlea/enzymology , Cochlea/metabolism , Cytoskeleton/metabolism , Deafness/genetics , Female , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , GTPase-Activating Proteins/genetics , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/physiology , Hearing Loss/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Stereocilia/enzymology
5.
Nat Commun ; 10(1): 4150, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31515474

ABSTRACT

Cisplatin is one of the most widely used chemotherapeutic drugs for the treatment of cancer. Unfortunately, one of its major side effects is permanent hearing loss. Here, we show that glutathione transferase α4 (GSTA4), a member of the Phase II detoxifying enzyme superfamily, mediates reduction of cisplatin ototoxicity by removing 4-hydroxynonenal (4-HNE) in the inner ears of female mice. Under cisplatin treatment, loss of Gsta4 results in more profound hearing loss in female mice compared to male mice. Cisplatin stimulates GSTA4 activity in the inner ear of female wild-type, but not male wild-type mice. In female Gsta4-/- mice, cisplatin treatment results in increased levels of 4-HNE in cochlear neurons compared to male Gsta4-/- mice. In CBA/CaJ mice, ovariectomy decreases mRNA expression of Gsta4, and the levels of GSTA4 protein in the inner ears. Thus, our findings suggest that GSTA4-dependent detoxification may play a role in estrogen-mediated neuroprotection.


Subject(s)
Cisplatin/adverse effects , Glutathione Transferase/metabolism , Ototoxicity/enzymology , Animals , Auditory Threshold/drug effects , Capillaries/pathology , Cochlea/enzymology , Cochlea/pathology , Cochlea/physiopathology , Crosses, Genetic , DNA Damage/genetics , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Gene Expression Regulation/drug effects , Glutathione Transferase/deficiency , Hearing Loss/complications , Hearing Loss/enzymology , Hearing Loss/physiopathology , Male , Mice, Inbred CBA , Ototoxicity/complications , Ototoxicity/pathology , Ototoxicity/physiopathology , Oxidative Stress/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spiral Ganglion/drug effects , Spiral Ganglion/pathology
6.
Hear Res ; 381: 107768, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31387070

ABSTRACT

The pupil dilation response is sensitive both to listening effort and the emotional significance of a task. We aimed to assess the influence of evaluative feedback on the pupil dilation response using a speech reception threshold (SRT) task. Besides the pupil dilation response, we acquired subjective ratings and two physiological biomarkers sensitive to stress: cortisol and alpha-amylase levels as determined in saliva samples. We included 34 participants with normal hearing (mean age = 52 years, age range 25-67 years) and 29 age-matched participants with mild-to-moderate hearing loss (mean age = 52 years, age range 23-64 years). Half of the participants performed a standard SRT test without feedback, and the other half performed an SRT test in which they did receive feedback and were urged to perform better. The SRT conditions targeted 50% and 71% correct reception of the sentences. Pupil size was recorded and saliva samples were obtained and participants rated their experience of the task. Participants with hearing loss performed more poorly on the SRT test than participants with normal hearing participants receiving feedback had better SRTs in the 71% intelligibility condition and higher peak pupil dilation in both intelligibility conditions than participants who did not receive feedback, irrespective of hearing status. Saliva cortisol level and alpha-amylase activity reflected the usual diurnal patterns but showed no effects of hearing status or feedback. Finally, participants who received feedback experienced more difficulties than those who did not receive feedback, irrespective of hearing status. This study underlines the importance of taking into account the influence of task instructions and feedback in a speech perception task as these factors may influence the experienced difficulties, listening effort, and task performance.


Subject(s)
Feedback, Psychological , Hearing Loss/psychology , Hearing , Hydrocortisone/metabolism , Persons With Hearing Impairments/psychology , Pupil , Saliva/enzymology , Speech Perception , alpha-Amylases/metabolism , Adult , Aged , Attention , Case-Control Studies , Female , Hearing Loss/diagnosis , Hearing Loss/enzymology , Hearing Loss/physiopathology , Humans , Male , Middle Aged , Speech Intelligibility , Speech Reception Threshold Test , Young Adult
7.
Hear Res ; 365: 1-15, 2018 08.
Article in English | MEDLINE | ID: mdl-29883832

ABSTRACT

As the COMT polymorphism is especially prominent in the prefrontal cortex and has been associated with auditory gating, we hypothesize that tinnitus patients with this polymorphism have altered activity in the ventromedial prefrontal/anterior cingulate areas that modulates the tinnitus percept. To test this, we recruited a total of 40 tinnitus subjects and 20 healthy controls for an EEG study. A comparison between tinnitus subjects and healthy controls and their frequency of being Val/Val genotype or Met carriers (including Val/Met and Met/Met genotype) shows no significant effect, suggesting that the distributions for the tinnitus and healthy groups are similar. Our results show that an interaction between the amount of hearing loss and the COMT Val158Met polymorphism can increase susceptibility to the clinical manifestation of tinnitus. We further demonstrate that the parahippocampus becomes involved in tinnitus in patients with hearing loss that are Met carriers. In these patients, the parahippocampus sends more tinnitus information to the pregenual anterior cingulate cortex and auditory cortex that is specifically related with increased loudness. At the same time, the pregenual anterior cingulate cortex, which normally functions as a gatekeeper, is not cancelling this auditory information, ultimately leading to increased tinnitus loudness.


Subject(s)
Brain/enzymology , Catechol O-Methyltransferase/genetics , Hearing Loss/genetics , Hearing/genetics , Polymorphism, Single Nucleotide , Tinnitus/genetics , Adult , Auditory Pathways/enzymology , Auditory Pathways/physiopathology , Auditory Perception/genetics , Brain/physiopathology , Case-Control Studies , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Hearing Loss/diagnosis , Hearing Loss/enzymology , Hearing Loss/physiopathology , Humans , Male , Middle Aged , Phenotype , Risk Factors , Severity of Illness Index , Tinnitus/diagnosis , Tinnitus/enzymology , Tinnitus/physiopathology
8.
Acta Otolaryngol ; 136(10): 991-8, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27192390

ABSTRACT

CONCLUSION: Nitric oxide synthase (NOS) II induction is a protective mechanism against age-related degeneration of the cochlea. OBJECTIVES: An induction of NOS II has been described in different inner ear pathologies. The objective was to examine the role of NOS II in age-related degeneration of the cochlea. METHODS: The hearing ability in adult and aging NOS II knockout mice (KO) and their wildtype (WT) littermates was explored via auditory brainstem response (ABR) measurements. Inner ear morphological differences were studied with scanning electron microscopy (SEM). Immunohistochemistry was used to examine the induction of NOS II in the inner ear of aging WT mice. Expression of nitrotyrosin, a marker protein for the reactive oxygen species peroxynitrite, was compared between KO and WT mice using immunohistochemistry. RESULTS: Adult KO mice exhibited a mild hearing impairment. WT mice showed an induction of NOS II after 6 months of age. Age-related hearing deterioration was accelerated in KO mice, which was accompanied by increased nitrotyrosin formation and outer hair cell loss.


Subject(s)
Aging/physiology , Cochlea/physiology , Hearing Loss/enzymology , Nitric Oxide Synthase Type II/physiology , Animals , Cochlea/pathology , Female , Hearing Loss/pathology , Immunohistochemistry , Mice, Knockout , Peroxynitrous Acid/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
9.
Sci Rep ; 6: 22573, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26934825

ABSTRACT

Noise-induced hearing loss (NIHL) is an important occupational disorder. However, the molecular mechanisms underlying NIHL have not been fully clarified; therefore, the condition lacks effective therapeutic methods. Cyclooxygenase-2 (Cox-2) is an inducible enzyme involved in the synthesis of prostaglandins, and has been implicated in many pathophysiological events, such as oxidative stress and inflammation. In this study, we investigated the possible role of Cox-2 in the mechanisms of NIHL and the therapeutic effect of the Cox-2 inhibitor NS398 on NIHL using a mouse model. We demonstrated that Cox-2 is constitutively expressed in the mouse cochlea, and its expression could be dramatically up-regulated by high levels of noise exposure. Furthermore, we demonstrated that pre-treatment with the Cox-2 inhibitor NS398 could inhibit Cox-2 expression during noise overstimulation; and could attenuate noise-induced hearing loss and hair cell damage. Our results suggest that Cox-2 is involved in the pathogenesis of NIHL; and pharmacological inhibition of Cox-2 has considerable therapeutic potential in NIHL.


Subject(s)
Cyclooxygenase 2/biosynthesis , Cyclooxygenase Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hearing Loss/drug therapy , Nitrobenzenes/pharmacology , Noise/adverse effects , Sulfonamides/pharmacology , Animals , Disease Models, Animal , Female , Hearing Loss/enzymology , Hearing Loss/etiology , Mice , Up-Regulation/drug effects
10.
Dis Model Mech ; 8(12): 1555-68, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26542706

ABSTRACT

Mitogen-activated protein kinase, MAP3K1, plays an important role in a number of cellular processes, including epithelial migration during eye organogenesis. In addition, studies in keratinocytes indicate that MAP3K1 signalling through JNK is important for actin stress fibre formation and cell migration. However, MAP3K1 can also act independently of JNK in the regulation of cell proliferation and apoptosis. We have identified a mouse mutant, goya, which exhibits the eyes-open-at-birth and microphthalmia phenotypes. In addition, these mice also have hearing loss. The goya mice carry a splice site mutation in the Map3k1 gene. We show that goya and kinase-deficient Map3k1 homozygotes initially develop supernumerary cochlear outer hair cells (OHCs) that subsequently degenerate, and a progressive profound hearing loss is observed by 9 weeks of age. Heterozygote mice also develop supernumerary OHCs, but no cellular degeneration or hearing loss is observed. MAP3K1 is expressed in a number of inner-ear cell types, including outer and inner hair cells, stria vascularis and spiral ganglion. Investigation of targets downstream of MAP3K1 identified an increase in p38 phosphorylation (Thr180/Tyr182) in multiple cochlear tissues. We also show that the extra OHCs do not arise from aberrant control of proliferation via p27KIP1. The identification of the goya mutant reveals a signalling molecule involved with hair-cell development and survival. Mammalian hair cells do not have the ability to regenerate after damage, which can lead to irreversible sensorineural hearing loss. Given the observed goya phenotype, and the many diverse cellular processes that MAP3K1 is known to act upon, further investigation of this model might help to elaborate upon the mechanisms underlying sensory hair cell specification, and pathways important for their survival. In addition, MAP3K1 is revealed as a new candidate gene for human sensorineural hearing loss.


Subject(s)
Hair Cells, Auditory, Outer/enzymology , Hair Cells, Auditory, Outer/pathology , MAP Kinase Kinase Kinase 1/genetics , Point Mutation/genetics , Animals , Base Sequence , Cell Proliferation , Cell Survival , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Hair Cells, Auditory, Outer/ultrastructure , Hearing , Hearing Loss/enzymology , Hearing Loss/pathology , Hearing Loss/physiopathology , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Phosphorylation , Vision, Ocular , beta-Galactosidase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Mol Med Rep ; 12(6): 7883-90, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26498835

ABSTRACT

Oxidative damage to mitochondrial DNA (mtDNA) and cell apoptosis are heavily implicated in aging. Our previous study established a mimetic rat model of aging in the cochleae using D­galactose (D­gal), and revealed that chronic injection of D­gal can increase oxidative stress and mtDNA common deletions (CD). The aim of the present study was to investigate the sources of reactive oxygen species and the occurrence of apoptosis in the cochleae of rats following 8 weeks of D­gal exposure. The results of the present study indicated that an elevated accumulation of the mtDNA CD and mitochondrial ultrastructural damage occurred in the cochleae of rats injected with D­gal for 8 weeks. In addition, the levels of 8­hydroxy­2­deoxyguanosine, NADPH oxidase (NOX) 3, P22phox and cleaved caspase 3, and the number of terminal deoxynucleotidyl transferase­mediated deoxyuridine triphosphate nick­end­labelling­positive cells were increased in the cochleae of D­gal­treated rats, compared with the controls. These findings suggested that nitric oxide synthase NOX3­associated oxidative stress may contribute to the accumulation of mtDNA mutations and activate a caspase 3­dependent apoptotic signalling pathway in the cochleae during aging. The present study also provided novel insights into the development of age­associated hearing loss, also termed presbycusis.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Cochlea/metabolism , Galactose/pharmacology , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Aging/metabolism , Aging/pathology , Animals , Cochlea/pathology , DNA, Mitochondrial , Hearing Loss/enzymology , Hearing Loss/pathology , In Situ Nick-End Labeling , Male , Rats , Rats, Sprague-Dawley , Sequence Deletion
12.
Nat Genet ; 47(3): 263-6, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25665007

ABSTRACT

Taking a genome-wide association study approach, we identified inherited genetic variations in ACYP2 associated with cisplatin-related ototoxicity (rs1872328: P = 3.9 × 10(-8), hazard ratio = 4.5) in 238 children with newly diagnosed brain tumors, with independent replication in 68 similarly treated children. The ACYP2 risk variant strongly predisposed these patients to precipitous hearing loss and was related to ototoxicity severity. These results point to new biology underlying the ototoxic effects of platinum agents.


Subject(s)
Acid Anhydride Hydrolases/genetics , Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Hearing Loss/chemically induced , Hearing Loss/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Child, Preschool , Clinical Trials as Topic , Female , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Hearing Loss/enzymology , Humans , Infant , Male , Polymorphism, Single Nucleotide , Acylphosphatase
13.
Otol Neurotol ; 35(5): e169-77, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24608370

ABSTRACT

HYPOTHESIS: To investigate whether curcumin may have in vivo protective effects against cisplatin ototoxicity by its direct scavenger activity and/or by curcumin-mediated upregulation of HO-1. BACKGROUND: Cisplatin-induced ototoxicity is a major dose-limiting side effect in anticancer chemotherapy. A protective approach to decrease cisplatin ototoxicity without compromising its therapeutic efficacy remains a critical goal for anticancer therapy. Recent evidences indicate that curcumin exhibits antioxidant, anti-inflammatory, and chemosensitizer activities. METHODS: In male adult Wistar rats, a curcumin dose of 200 mg/kg, selected from a dose-response curve, was injected 1 hour before cisplatin administration and once daily for the following 3 days. A single dose of cisplatin (16 mg/kg) was administered intraperitoneally. Rats were divided as follows: 1) control, 2) curcumin control, 3) vehicle control, 4) cisplatin, 5) cisplatin+ vehicle, and 6) curcumin+cisplatin. ABRs were measured before and at Days 3 and 5 after cisplatin administration. Rhodamine-phalloidin staining, 4-hydroxy-2-nonenal and heme-oxigenase-1 immunostainings, and Western blot analyses were performed to assess and quantify OHC loss, lipid peroxidation, and the endogenous response to cisplatin-induced damage and to curcumin protection. RESULTS: Curcumin treatment attenuated hearing loss induced by cisplatin, increased OHC survival, decreased 4-HNE expression, and increased HO-1 expression. CONCLUSION: This preclinical study demonstrates that systemic curcumin attenuates ototoxicity and provides molecular evidence for a role of HO-1 as an additional mediator in attenuating cisplatin-induced damage.


Subject(s)
Curcumin/therapeutic use , Evoked Potentials, Auditory, Brain Stem/drug effects , Hearing Loss/drug therapy , Heme Oxygenase-1/metabolism , Animals , Cisplatin , Curcumin/pharmacology , Hearing Loss/chemically induced , Hearing Loss/enzymology , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/enzymology , Male , Rats , Rats, Wistar
14.
J Med Genet ; 51(4): 275-82, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24556084

ABSTRACT

BACKGROUND: Congenital muscular dystrophies (CMD) with hypoglycosylation of α-dystroglycan are clinically and genetically heterogeneous disorders that are often associated with brain malformations and eye defects. Presently, 16 proteins are known whose dysfunction impedes glycosylation of α-dystroglycan and leads to secondary dystroglycanopathy. OBJECTIVE: To identify the cause of CMD with secondary merosin deficiency, hypomyelination and intellectual disability in two siblings from a consanguineous family. METHODS: Autozygosity mapping followed by whole exome sequencing and immunochemistry were used to discover and verify a new genetic defect in two siblings with CMD. RESULTS: We identified a homozygous missense mutation (c.325C>T, p.Q109*) in protein O-mannosyl kinase (POMK) that encodes a glycosylation-specific kinase (SGK196) required for function of the dystroglycan complex. The protein was absent from skeletal muscle and skin fibroblasts of the patients. In patient muscle, ß-dystroglycan was normally expressed at the sarcolemma, while α-dystroglycan failed to do so. Further, we detected co-localisation of POMK with desmin at the costameres in healthy muscle, and a substantial loss of desmin from the patient muscle. CONCLUSIONS: Homozygous truncating mutations in POMK lead to CMD with secondary merosin deficiency, hypomyelination and intellectual disability. Loss of desmin suggests that failure of proper α-dystroglycan glycosylation impedes the binding to extracellular matrix proteins and also affects the cytoskeleton.


Subject(s)
Hearing Loss/complications , Intellectual Disability/complications , Laminin/deficiency , Muscular Dystrophies/congenital , Mutation/genetics , Myelin Sheath/pathology , Protein Kinases/genetics , Adolescent , Base Sequence , Child , Child, Preschool , DNA Mutational Analysis , Family , Female , Hearing Loss/enzymology , Hearing Loss/genetics , Humans , Immunohistochemistry , Infant , Infant, Newborn , Intellectual Disability/enzymology , Intellectual Disability/genetics , Male , Molecular Sequence Data , Muscle, Skeletal/pathology , Muscular Dystrophies/complications , Muscular Dystrophies/enzymology , Muscular Dystrophies/genetics , Pedigree , Young Adult
15.
Hum Mol Genet ; 23(12): 3289-98, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24482543

ABSTRACT

Cyclic AMP (cAMP) production, which is important for mechanotransduction within the inner ear, is catalyzed by adenylate cyclases (AC). However, knowledge of the role of ACs in hearing is limited. Previously, a novel autosomal recessive non-syndromic hearing impairment locus DFNB44 was mapped to chromosome 7p14.1-q11.22 in a consanguineous family from Pakistan. Through whole-exome sequencing of DNA samples from hearing-impaired family members, a nonsense mutation c.3112C>T (p.Arg1038*) within adenylate cyclase 1 (ADCY1) was identified. This stop-gained mutation segregated with hearing impairment within the family and was not identified in ethnically matched controls or within variant databases. This mutation is predicted to cause the loss of 82 amino acids from the carboxyl tail, including highly conserved residues within the catalytic domain, plus a calmodulin-stimulation defect, both of which are expected to decrease enzymatic efficiency. Individuals who are homozygous for this mutation had symmetric, mild-to-moderate mixed hearing impairment. Zebrafish adcy1b morphants had no FM1-43 dye uptake and lacked startle response, indicating hair cell dysfunction and gross hearing impairment. In the mouse, Adcy1 expression was observed throughout inner ear development and maturation. ADCY1 was localized to the cytoplasm of supporting cells and hair cells of the cochlea and vestibule and also to cochlear hair cell nuclei and stereocilia. Ex vivo studies in COS-7 cells suggest that the carboxyl tail of ADCY1 is essential for localization to actin-based microvilli. These results demonstrate that ADCY1 has an evolutionarily conserved role in hearing and that cAMP signaling is important to hair cell function within the inner ear.


Subject(s)
Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Cyclic AMP/metabolism , Ear, Inner/metabolism , Hair Cells, Auditory/metabolism , Hearing Loss/pathology , Adenylyl Cyclases/chemistry , Animals , COS Cells , Chlorocebus aethiops , Codon, Nonsense , Cytoplasm/metabolism , Ear, Inner/growth & development , Female , Hearing Loss/enzymology , Humans , Labyrinth Supporting Cells/metabolism , Male , Mice , Zebrafish/genetics
16.
Hum Mol Genet ; 23(6): 1591-601, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24191262

ABSTRACT

Methionine sulfoxide reductase B3 (MsrB3) is a protein repair enzyme that specifically reduces methionine-R-sulfoxide to methionine. A recent genetic study showed that the MSRB3 gene is associated with autosomal recessive hearing loss in human deafness DFNB74. However, the precise role of MSRB3 in the auditory system and the pathogenesis of hearing loss have not yet been determined. This work is the first to generate MsrB3 knockout mice to elucidate the possible pathological mechanisms of hearing loss observed in DFNB74 patients. We found that homozygous MsrB3(-/-) mice were profoundly deaf and had largely unaffected vestibular function, whereas heterozygous MsrB3(+/-) mice exhibited normal hearing similar to that of wild-type mice. The MsrB3 protein is expressed in the sensory epithelia of the cochlear and vestibular tissues, beginning at E15.5 and E13.5, respectively. Interestingly, MsrB3 is densely localized at the base of stereocilia on the apical surface of auditory hair cells. MsrB3 deficiency led to progressive degeneration of stereociliary bundles starting at P8, followed by a loss of hair cells, resulting in profound deafness in MsrB3(-/-) mice. The hair cell loss appeared to be mediated by apoptotic cell death, which was measured using TUNEL and caspase 3 immunocytochemistry. Taken together, our data suggest that MsrB3 plays an essential role in maintaining the integrity of hair cells, possibly explaining the pathogenesis of DFNB74 deafness in humans caused by MSRB3 deficiency.


Subject(s)
Cochlea/pathology , Hearing Loss/genetics , Hearing Loss/pathology , Methionine Sulfoxide Reductases/genetics , Stereocilia/pathology , Animals , Apoptosis , Disease Models, Animal , Gene Expression Regulation, Developmental , Hair Cells, Auditory/pathology , Hearing Loss/enzymology , Humans , Methionine Sulfoxide Reductases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Stereocilia/metabolism
17.
Oxid Med Cell Longev ; 2013: 325702, 2013.
Article in English | MEDLINE | ID: mdl-23936608

ABSTRACT

Age-related hearing loss (AHL) reduces the quality of life for many elderly individuals. Manganese superoxide dismutase (Mn-SOD), one of the antioxidant enzymes acting within the mitochondria, plays a crucial role in scavenging reactive oxygen species (ROS). To determine whether reduction in Mn-SOD accelerates AHL, we evaluated auditory function in Mn-SOD heterozygous knockout (HET) mice and their littermate wild-type (WT) C57BL/6 mice by means of auditory brainstem response (ABR). Mean ABR thresholds were significantly increased at 16 months when compared to those at 4 months in both WT and HET mice, but they did not significantly differ between them at either age. The extent of hair cell loss, spiral ganglion cell density, and thickness of the stria vascularis also did not differ between WT and HET mice at either age. At 16 months, immunoreactivity of 8-hydroxydeoxyguanosine was significantly greater in the SGC and SV in HET mice compared to WT mice, but that of 4-hydroxynonenal did not differ between them. These findings suggest that, although decrease of Mn-SOD by half may increase oxidative stress in the cochlea to some extent, it may not be sufficient to accelerate age-related cochlear damage under physiological aging process.


Subject(s)
Aging/physiology , Hearing Loss/enzymology , Superoxide Dismutase/deficiency , Animals , Hearing Loss/genetics , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics
18.
Am J Hum Genet ; 93(1): 132-40, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23768514

ABSTRACT

Previously, DFNB89, a locus associated with autosomal-recessive nonsyndromic hearing impairment (ARNSHI), was mapped to chromosomal region 16q21-q23.2 in three unrelated, consanguineous Pakistani families. Through whole-exome sequencing of a hearing-impaired individual from each family, missense mutations were identified at highly conserved residues of lysyl-tRNA synthetase (KARS): the c.1129G>A (p.Asp377Asn) variant was found in one family, and the c.517T>C (p.Tyr173His) variant was found in the other two families. Both variants were predicted to be damaging by multiple bioinformatics tools. The two variants both segregated with the nonsyndromic-hearing-impairment phenotype within the three families, and neither mutation was identified in ethnically matched controls or within variant databases. Individuals homozygous for KARS mutations had symmetric, severe hearing impairment across all frequencies but did not show evidence of auditory or limb neuropathy. It has been demonstrated that KARS is expressed in hair cells of zebrafish, chickens, and mice. Moreover, KARS has strong localization to the spiral ligament region of the cochlea, as well as to Deiters' cells, the sulcus epithelium, the basilar membrane, and the surface of the spiral limbus. It is hypothesized that KARS variants affect aminoacylation in inner-ear cells by interfering with binding activity to tRNA or p38 and with tetramer formation. The identification of rare KARS variants in ARNSHI-affected families defines a gene that is associated with ARNSHI.


Subject(s)
Hearing Loss/enzymology , Hearing Loss/genetics , Lysine-tRNA Ligase/genetics , Mutation, Missense , Animals , Case-Control Studies , Chickens , Cochlea/metabolism , Cochlea/pathology , Computational Biology/methods , Consanguinity , Female , Genetic Linkage , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Haplotypes , Homozygote , Humans , Lysine-tRNA Ligase/metabolism , Male , Mice , Pedigree , Transfer RNA Aminoacylation , Zebrafish/embryology , Zebrafish/metabolism
19.
Dis Model Mech ; 6(2): 434-42, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23065636

ABSTRACT

Mutations in the ATP6V0A4 gene lead to autosomal recessive distal renal tubular acidosis in patients, who often show sensorineural hearing impairment. A first Atp6v0a4 knockout mouse model that recapitulates the loss of H(+)-ATPase function seen in humans has been generated and recently reported (Norgett et al., 2012). Here, we present the first detailed analysis of the structure and function of the auditory system in Atp6v0a4(-/-) knockout mice. Measurements of the auditory brainstem response (ABR) showed significantly elevated thresholds in homozygous mutant mice, which indicate severe hearing impairment. Heterozygote thresholds were normal. Analysis of paint-filled inner ears and sections from E16.5 embryos revealed a marked expansion of cochlear and endolymphatic ducts in Atp6v0a4(-/-) mice. A regulatory link between Atp6v0a4, Foxi1 and Pds has been reported and we found that the endolymphatic sac of Atp6v0a4(-/-) mice expresses both Foxi1 and Pds, which suggests a downstream position of Atp6v0a4. These mutants also showed a lack of endocochlear potential, suggesting a functional defect of the stria vascularis on the lateral wall of the cochlear duct. However, the main K(+) channels involved in the generation of endocochlear potential, Kcnj10 and Kcnq1, are strongly expressed in Atp6v0a4(-/-) mice. Our results lead to a better understanding of the role of this proton pump in hearing function.


Subject(s)
Ear, Inner/enzymology , Ear, Inner/pathology , Endolymph/enzymology , Hearing Loss/enzymology , Hearing Loss/pathology , Protein Subunits/deficiency , Proton-Translocating ATPases/deficiency , Animals , Animals, Newborn , Anion Transport Proteins/metabolism , Ear, Inner/physiopathology , Endolymphatic Sac/pathology , Endolymphatic Sac/physiopathology , Epithelium/metabolism , Epithelium/pathology , Evoked Potentials, Auditory , Forkhead Transcription Factors/metabolism , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Hair Cells, Auditory, Outer/ultrastructure , Hearing Loss/physiopathology , Humans , KCNQ1 Potassium Channel/metabolism , Mice , Mice, Knockout , Mutation/genetics , Phenotype , Potassium Channels, Inwardly Rectifying/metabolism , Protein Subunits/metabolism , Proton-Translocating ATPases/metabolism , Stria Vascularis/metabolism , Stria Vascularis/pathology , Sulfate Transporters , Vacuolar Proton-Translocating ATPases
20.
Int J Audiol ; 52(1): 23-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23190330

ABSTRACT

OBJECTIVE: The purpose of this review was to evaluate the current literature on phosphoribosylpyrophosphate synthetase 1 (PRPS1)-related diseases and their consequences on hearing function. DESIGN: A literature search of peer-reviewed, published journal articles was conducted in online bibliographic databases. STUDY SAMPLE: Three databases for medical research were included in this review. RESULTS: Mutations in PRPS1 are associated with a spectrum of non-syndromic to syndromic hearing loss. Hearing loss in male patients with PRPS1 mutations is bilateral, moderate to profound, and can be prelingual or postlingual, progressive or non-progressive. Audiogram shapes associated with PRPS1 deafness are usually residual and flat. Female carriers can have unilateral or bilateral hearing impairment. Gain of function mutations in PRPS1 cause a superactivity of the PRS-I protein whereas the loss-of-function mutations result in X-linked nonsyndromic sensorineural deafness type 2 (DFN2), or in syndromic deafness including Arts syndrome and X-linked Charcot-Marie-Tooth disease-5 (CMTX5). CONCLUSIONS: Lower residual activity in PRS-I leads to a more severe clinical manifestation. Clinical and molecular findings suggest that the four PRPS1 disorders discovered to date belong to the same disease spectrum. Dietary supplementation with S-adenosylmethionine (SAM) appeared to alleviate the symptoms of Arts syndrome patients, suggesting that SAM could compensate for PRS-I deficiency.


Subject(s)
Hearing Loss/genetics , Hearing/genetics , Mutation , Ribose-Phosphate Pyrophosphokinase/genetics , Dietary Supplements , Female , Genetic Predisposition to Disease , Hearing Loss/diagnosis , Hearing Loss/drug therapy , Hearing Loss/enzymology , Hearing Loss/physiopathology , Heredity , Humans , Male , Phenotype , S-Adenosylmethionine/therapeutic use , Severity of Illness Index , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL